Skip to main content
Journal of Bacteriology logoLink to Journal of Bacteriology
. 1995 Nov;177(22):6330–6337. doi: 10.1128/jb.177.22.6330-6337.1995

Pseudomonas aeruginosa sodA and sodB mutants defective in manganese- and iron-cofactored superoxide dismutase activity demonstrate the importance of the iron-cofactored form in aerobic metabolism.

D J Hassett 1, H P Schweizer 1, D E Ohman 1
PMCID: PMC177481  PMID: 7592406

Abstract

The consumption of molecular oxygen by Pseudomonas aeruginosa can lead to the production of reduced oxygen species, including superoxide, hydrogen peroxide, and the hydroxyl radical. As a first line of defense against potentially toxic levels of endogenous superoxide, P. aeruginosa possesses an iron- and manganese-cofactored superoxide dismutase (SOD) to limit the damage evoked by this radical. In this study, we have generated mutants which possess an interrupted sodA (encoding manganese SOD) or sodB (encoding iron SOD) gene and a sodA sodB double mutant. Mutagenesis of sodA did not significantly alter the aerobic growth rate in rich medium (Luria broth) or in glucose minimal medium in comparison with that of wild-type bacteria. In addition, total SOD activity in the sodA mutant was decreased only 15% relative to that of wild-type bacteria. In contrast, sodB mutants grew much more slowly than the sodA mutant or wild-type bacteria in both media, and sodB mutants possessed only 13% of the SOD activity of wild-type bacteria. There was also a progressive decrease in catalase activity in each of the mutants, with the sodA sodB double mutant possessing only 40% of the activity of wild-type bacteria. The sodA sodB double mutant grew very slowly in rich medium and required approximately 48 h to attain saturated growth in minimal medium. There was no difference in growth of either strain under anaerobic conditions. Accordingly, the sodB but not the sodA mutant demonstrated marked sensitivity to paraquat, a superoxide-generating agent. P. aeuroginosa synthesizes a blue, superoxide-generating antibiotic similar to paraquat in redox properties which is called pyocyanin, the synthesis of which is accompanied by increased iron SOD and catalase activities (D.J. Hassett, L. Charniga, K. A. Bean, D. E. Ohman, and M. S. Cohen, Infect. Immun. 60:328-336, 1992). Pyocyanin production was completely abolished in the sodB and sodA sodB mutants and was decreased approximately 57% in sodA mutants relative to that of the wild-type organism. Furthermore, the addition of sublethal concentrations of paraquat to wild-type bacteria caused a concentration-dependent decrease in pyocyanin production, suggesting that part of the pyocyanin biosynthetic cascade is inhibited by superoxide. These results suggest that iron SOD is more important than manganese SOD for aerobic growth, resistance to paraquat, and optimal pyocyanin biosynthesis in P. aeruginosa.

Full Text

The Full Text of this article is available as a PDF (424.1 KB).

Selected References

These references are in PubMed. This may not be the complete list of references from this article.

  1. Archibald F. S., Duong M. N. Superoxide dismutase and oxygen toxicity defenses in the genus Neisseria. Infect Immun. 1986 Feb;51(2):631–641. doi: 10.1128/iai.51.2.631-641.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Archibald F. S., Fridovich I. Manganese and defenses against oxygen toxicity in Lactobacillus plantarum. J Bacteriol. 1981 Jan;145(1):442–451. doi: 10.1128/jb.145.1.442-451.1981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. BEERS R. F., Jr, SIZER I. W. A spectrophotometric method for measuring the breakdown of hydrogen peroxide by catalase. J Biol Chem. 1952 Mar;195(1):133–140. [PubMed] [Google Scholar]
  4. Benov L. T., Fridovich I. Escherichia coli expresses a copper- and zinc-containing superoxide dismutase. J Biol Chem. 1994 Oct 14;269(41):25310–25314. [PubMed] [Google Scholar]
  5. Beyer W., Imlay J., Fridovich I. Superoxide dismutases. Prog Nucleic Acid Res Mol Biol. 1991;40:221–253. doi: 10.1016/s0079-6603(08)60843-0. [DOI] [PubMed] [Google Scholar]
  6. Bradford M. M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976 May 7;72:248–254. doi: 10.1006/abio.1976.9999. [DOI] [PubMed] [Google Scholar]
  7. Carlioz A., Touati D. Isolation of superoxide dismutase mutants in Escherichia coli: is superoxide dismutase necessary for aerobic life? EMBO J. 1986 Mar;5(3):623–630. doi: 10.1002/j.1460-2075.1986.tb04256.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Clare D. A., Duong M. N., Darr D., Archibald F., Fridovich I. Effects of molecular oxygen on detection of superoxide radical with nitroblue tetrazolium and on activity stains for catalase. Anal Biochem. 1984 Aug 1;140(2):532–537. doi: 10.1016/0003-2697(84)90204-5. [DOI] [PubMed] [Google Scholar]
  9. Cox C. D. Role of pyocyanin in the acquisition of iron from transferrin. Infect Immun. 1986 Apr;52(1):263–270. doi: 10.1128/iai.52.1.263-270.1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Essar D. W., Eberly L., Hadero A., Crawford I. P. Identification and characterization of genes for a second anthranilate synthase in Pseudomonas aeruginosa: interchangeability of the two anthranilate synthases and evolutionary implications. J Bacteriol. 1990 Feb;172(2):884–900. doi: 10.1128/jb.172.2.884-900.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Farr S. B., D'Ari R., Touati D. Oxygen-dependent mutagenesis in Escherichia coli lacking superoxide dismutase. Proc Natl Acad Sci U S A. 1986 Nov;83(21):8268–8272. doi: 10.1073/pnas.83.21.8268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Farr S. B., Touati D., Kogoma T. Effects of oxygen stress on membrane functions in Escherichia coli: role of HPI catalase. J Bacteriol. 1988 Apr;170(4):1837–1842. doi: 10.1128/jb.170.4.1837-1842.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Fridovich I. Superoxide dismutases: regularities and irregularities. Harvey Lect. 1983 1984;79:51–75. [PubMed] [Google Scholar]
  14. Gardner P. R., Fridovich I. Superoxide sensitivity of the Escherichia coli 6-phosphogluconate dehydratase. J Biol Chem. 1991 Jan 25;266(3):1478–1483. [PubMed] [Google Scholar]
  15. Gardner P. R., Fridovich I. Superoxide sensitivity of the Escherichia coli aconitase. J Biol Chem. 1991 Oct 15;266(29):19328–19333. [PubMed] [Google Scholar]
  16. Haas D., Holloway B. W., Schamböck A., Leisinger T. The genetic organization of arginine biosynthesis in Pseudomonas aeruginosa. Mol Gen Genet. 1977 Jul 7;154(1):7–22. doi: 10.1007/BF00265571. [DOI] [PubMed] [Google Scholar]
  17. Hassan H. M., Fridovich I. Enzymatic defenses against the toxicity of oxygen and of streptonigrin in Escherichia coli. J Bacteriol. 1977 Mar;129(3):1574–1583. doi: 10.1128/jb.129.3.1574-1583.1977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Hassan H. M., Fridovich I. Mechanism of the antibiotic action pyocyanine. J Bacteriol. 1980 Jan;141(1):156–163. doi: 10.1128/jb.141.1.156-163.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Hassan H. M., Fridovich I. Paraquat and Escherichia coli. Mechanism of production of extracellular superoxide radical. J Biol Chem. 1979 Nov 10;254(21):10846–10852. [PubMed] [Google Scholar]
  20. Hassan H. M., Fridovich I. Regulation of the synthesis of superoxide dismutase in Escherichia coli. Induction by methyl viologen. J Biol Chem. 1977 Nov 10;252(21):7667–7672. [PubMed] [Google Scholar]
  21. Hassett D. J., Charniga L., Bean K., Ohman D. E., Cohen M. S. Response of Pseudomonas aeruginosa to pyocyanin: mechanisms of resistance, antioxidant defenses, and demonstration of a manganese-cofactored superoxide dismutase. Infect Immun. 1992 Feb;60(2):328–336. doi: 10.1128/iai.60.2.328-336.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Hassett D. J., Woodruff W. A., Wozniak D. J., Vasil M. L., Cohen M. S., Ohman D. E. Cloning and characterization of the Pseudomonas aeruginosa sodA and sodB genes encoding manganese- and iron-cofactored superoxide dismutase: demonstration of increased manganese superoxide dismutase activity in alginate-producing bacteria. J Bacteriol. 1993 Dec;175(23):7658–7665. doi: 10.1128/jb.175.23.7658-7665.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Herbert S. K., Samson G., Fork D. C., Laudenbach D. E. Characterization of damage to photosystems I and II in a cyanobacterium lacking detectable iron superoxide dismutase activity. Proc Natl Acad Sci U S A. 1992 Sep 15;89(18):8716–8720. doi: 10.1073/pnas.89.18.8716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Imlay J. A., Fridovich I. Suppression of oxidative envelope damage by pseudoreversion of a superoxide dismutase-deficient mutant of Escherichia coli. J Bacteriol. 1992 Feb;174(3):953–961. doi: 10.1128/jb.174.3.953-961.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Klug D., Rabani J., Fridovich I. A direct demonstration of the catalytic action of superoxide dismutase through the use of pulse radiolysis. J Biol Chem. 1972 Aug 10;247(15):4839–4842. [PubMed] [Google Scholar]
  26. Kono Y., Fridovich I. Superoxide radical inhibits catalase. J Biol Chem. 1982 May 25;257(10):5751–5754. [PubMed] [Google Scholar]
  27. Kuo C. F., Mashino T., Fridovich I. alpha, beta-Dihydroxyisovalerate dehydratase. A superoxide-sensitive enzyme. J Biol Chem. 1987 Apr 5;262(10):4724–4727. [PubMed] [Google Scholar]
  28. Liochev S. I., Fridovich I. Fumarase C, the stable fumarase of Escherichia coli, is controlled by the soxRS regulon. Proc Natl Acad Sci U S A. 1992 Jul 1;89(13):5892–5896. doi: 10.1073/pnas.89.13.5892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. McCord J. M., Fridovich I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J Biol Chem. 1969 Nov 25;244(22):6049–6055. [PubMed] [Google Scholar]
  30. Meade H. M., Long S. R., Ruvkun G. B., Brown S. E., Ausubel F. M. Physical and genetic characterization of symbiotic and auxotrophic mutants of Rhizobium meliloti induced by transposon Tn5 mutagenesis. J Bacteriol. 1982 Jan;149(1):114–122. doi: 10.1128/jb.149.1.114-122.1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Nakayama K. Rapid viability loss on exposure to air in a superoxide dismutase-deficient mutant of Porphyromonas gingivalis. J Bacteriol. 1994 Apr;176(7):1939–1943. doi: 10.1128/jb.176.7.1939-1943.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Sadosky A. B., Wilson J. W., Steinman H. M., Shuman H. A. The iron superoxide dismutase of Legionella pneumophila is essential for viability. J Bacteriol. 1994 Jun;176(12):3790–3799. doi: 10.1128/jb.176.12.3790-3799.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Schweizer H. D. Small broad-host-range gentamycin resistance gene cassettes for site-specific insertion and deletion mutagenesis. Biotechniques. 1993 Nov;15(5):831–834. [PubMed] [Google Scholar]
  34. Schweizer H. P. Allelic exchange in Pseudomonas aeruginosa using novel ColE1-type vectors and a family of cassettes containing a portable oriT and the counter-selectable Bacillus subtilis sacB marker. Mol Microbiol. 1992 May;6(9):1195–1204. doi: 10.1111/j.1365-2958.1992.tb01558.x. [DOI] [PubMed] [Google Scholar]
  35. Simon R., O'Connell M., Labes M., Pühler A. Plasmid vectors for the genetic analysis and manipulation of rhizobia and other gram-negative bacteria. Methods Enzymol. 1986;118:640–659. doi: 10.1016/0076-6879(86)18106-7. [DOI] [PubMed] [Google Scholar]
  36. Southern E. M. Detection of specific sequences among DNA fragments separated by gel electrophoresis. J Mol Biol. 1975 Nov 5;98(3):503–517. doi: 10.1016/s0022-2836(75)80083-0. [DOI] [PubMed] [Google Scholar]
  37. Steinman H. M. Bacteriocuprein superoxide dismutases in pseudomonads. J Bacteriol. 1985 Jun;162(3):1255–1260. doi: 10.1128/jb.162.3.1255-1260.1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. VOGEL H. J., BONNER D. M. Acetylornithinase of Escherichia coli: partial purification and some properties. J Biol Chem. 1956 Jan;218(1):97–106. [PubMed] [Google Scholar]
  39. Wieslander L. A simple method to recover intact high molecular weight RNA and DNA after electrophoretic separation in low gelling temperature agarose gels. Anal Biochem. 1979 Oct 1;98(2):305–309. doi: 10.1016/0003-2697(79)90145-3. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Bacteriology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES